Review

Secretion, migration and adhesion as key processes in the therapeutic activity of mesenchymal stem cells*

Marta Kot, Aleksandra Musiał-Wysocka, Małgorzata Lasota, Aleksandra Ulman
and Marcin Majka

Jagiellonian University, Medical College, Department of Transplantation, Kraków, Poland

The MSCs are immature cells that can be found in numerous different tissue types. In recent years, they have gained considerable attention, particularly with regard to their regenerative properties. Due to their paracrine activity, ability to migrate, adhesion and homing, MSCs currently appear to be the most relevant for therapeutic use. Numerous bioactive molecules secreted by MSCs exert paracrine effects and modulate many physiological processes, such as angiogenesis, immunomodulation and neuroprotection. Cell-cell communication may be also mediated by extracellular vesicles released from the cells. Due to these properties, MSCs have been widely studied for evaluation of their therapeutic benefits expected in the clinical applications. For effective tissue regeneration, transplanted MSCs have to exit the circulation and locate at the site of damage, which is possible because of their ability to migrate, adhere and engraft at the target site. Accumulating evidence suggests that MSCs recruitment from remote sites is similar to leukocytes’ migration. All of these biological features make MSCs highly investigated stem cells and the most commonly used cells in regenerative medicine. Since environmental factors affect the MSCs behavior, we discuss importance of oxygen concentration as a one of the key factors affecting MSCs properties.

Key words: mesenchymal stem cells (MSCs), secretion, adhesion, migration, homing, cell therapy

Received: 21 October, 2019; revised: 17 December, 2019; accepted: 19 December, 2019; available on-line: 28 December, 2019

e-mail: mmajka@cm-uj.krakow.pl (MM); marta.kot@uj.edu.pl (MK)

*Acknowlkedgements of Financial Support:

The costs of the article published as a part of the 44th FEBS Congress Kraków 2019 – From molecules to living systems block are financed by the Ministry of Science and Higher Education of the Republic of Poland (Contract 805/P-DUN/2019).

This work was supported by a research grant (STRATEGMED2/265761/10/NCBR/2015) from the National Center for Research and Development.

Abbreviations: MSCs, mesenchymal stem cells; EVs, extracellular vesicles; SDF-1α, cell-derived factor-1 alpha chemokine; MMP1, matrix metalloproteinase-1; ECM, extracellular matrix; VEGF, vascular endothelial growth factor; HIF-1α, hypoxia-inducible factor 1 alpha

Introduction

Mesenchymal stem cells (MSCs) are self-renewing, multipotent cells that can be isolated from nearly every tissue type (bone marrow, adipose tissue, umbilical cord, peripheral blood, placenta) (Zvaifler et al., 2000; Zuk et al., 2001; Najar et al., 2010; Fei et al., 2013; Heidari et al., 2013; Meier et al., 2013; Musialek et al., 2015; Musiał-Wysocka et al., 2019a, Musiał-Wysocka et al., 2019b). A wide variety of tissue sources implies high heterogeneity of the isolated populations. Thus, MSCs are identified according to criteria established by ISTC in 2006, which have been summarized by Dominici and others (Dominici et al., 2006). An important property of MSCs is their ability to secrete numerous factors involved in different processes that favor tissue remodeling, such as angiogenesis, immunomodulation or neuroprotection. All of these biological features, combined with ease of culture expansion, make MSCs highly investigated stem cells and the most commonly used cells in regenerative medicine (Lee et al., 2010; Wang et al., 2012; Wei et al., 2013; Hare et al., 2017). It should be noted that despite general unified criteria outlined for MSCs, the cells isolated from different tissues may vary in terms of their potential to differentiate, proliferate or their profile of secreted factors. MSCs retain properties of the microenvironment from which they are derived in vivo (Mrozik et al., 2010).

The available data allow to distinguish some mechanisms that are responsible for the therapeutic potential of mesenchymal stem cells. Positive effect of exogenously injected MSCs can be a result of both, the cell-cell interaction and the paracrine activity, such as:

One of the most important challenges of the current clinical use of MSCs is the comprehensive understanding of their biology, in particular the mechanism underlying the basis of their ability to regenerate damaged tissues. Despite accumulating evidence, the nature and functions of MSCs are not fully understood. Taking into consideration all MSCs properties known so far, it is difficult to indicate only one of them that might be responsible for the therapeutic abilities of these cells. In this paper, we discuss the current understanding of MSCs properties that are inseparably connected with their regenerative potential, such as the secretory activity and ability to migrate, adhere and home. It should be noted that the in vitro culture conditions under which the cells are examined differ from the in vivo environment, thus we have also considered the importance of oxygen concentration in MSCs investigations.

The MSCs’ paracrine effect: Secretome and MSCs-released vesicles

MSCs secretome

The ability to secrete a variety of bioactive factors (cytokines, chemokines, and growth factors) is one of the key activities of MSCs. The released molecules are involved in interactions between MSCs or the surrounding microenvironment.

The spectrum of compounds produced and released by MSCs, generally referred to as the MSCs secretome, can be divided into several panels of factors involved in various biological processes, i.e. angiogenesis, immunomodulation, neuroprotection, proliferation, migration, and chemotaxis (Fig. 1).

MSCs are tested in many preclinical and clinical studies as a promising approach for various therapies. They gained special attention in tissue regeneration, where the paracrine effect of MSCs is of particular interest. The ability to secrete biologically active molecules that can affect nearby cells is considered as key MSCs activity. It depends on the histological source of cells, the donor age and impact of the surrounding microenvironment. MSCs secretome composition can vary significantly (Doorn et al., 2012). It is abundant in growth factors, cytokines and chemokines, therefore making MSCs a perfect source of a conditioned media for many studies concerning cell-free therapy (Praveen et al., 2019). Based on the composition and biological effects, we can divide the MSCs secretome into immunomodulating, neuroprotective, tissue remodeling and angiogenic.

Immunomodulation

MSCs’ properties of regulating the immune system are based not only on the particular surface molecules but also on secretion of many soluble factors. MSCs secrete both, the anti-inflammatory cytokines such as the transforming growth factor β1 (TGF-β1), interleukin 13 (IL-13), and pro-inflammatory cytokines such as IL1b, IL6, IL8 and IL9 (Vizoso et al., 2017). The importance of this equilibrium is presented with the results from a study with a mouse model of diabetics. It was shown that after application of conditioned media from the adipose tissue, the MSCs balance in the pro- and anti-inflammatory factors was restored. The IL-10 level was decreased, whereas the IL-1β, IL-6 and TNF-α levels were elevated in the dorsal root ganglia and the spinal cord (Brini et al., 2017).

Neuroprotection

The nervous system is characterized by a low regeneration potential. Therefore, studies that associate MSCs with improved nervous system recovery are substantial for the future therapies. In the beginning those functions were correlated with the differentiation capacity of MSCs, but recent studies reveal the importance of the MSCs secretome in which we can find many neuroprotective and neuroregulatory factors (Teixeira et al., 2013). Crigler and others (Crigler et al., 2006) discovered transcripts coding a brain-derived neurotrophic factor (BDNF) and nerve growth factor (β-NGF), after analyzing MSCs cDNA library. What is more, they correlate the nerve survival and outgrowth improvement with BDNF activity from co-culture experiments (Crigler et al., 2006). Further studies reveal that BDNF is a key factor secreted by MSCs from Wharton’s Jelly of the umbilical cord (WJ-MSCs), which exclusively mediates axonal elongation in the rat cortical and hippocampal neurons (Martins et al., 2017).

Angiogenic

MSCs are widely studied in research concerning ischemic recovery. Angiogenic properties that promote sprouting of new blood vessels from preexisting ones seem to be a crucial regenerative feature of MSCs. Many studies confirm this activity owing to production of a variety of factors, such as the vascular endothelial growth factor (VEGF-A), angiopoietins (ANGPTs), insulin-like growth factor-1 (IGF-1) or hepatocyte growth factor (HGF). VEGF is indicated as one of the most abundant proteins secreted by MSCs (Ferreira et al., 2018). The cell culture conditions seem to be crucial for the angiogenic secretome properties. It was shown by Oskowitz et al. (2011) that a serum deprivation condition results in an increased expression of angiogenic factors, such as the VEGF-A, ANGPTs, IGF-1, and HGF. The angiogenic potential of the secretome was confirmed using an ex vivo rat aortic ring assay where media from serum deprived MSCs resulted in significantly longer vascular sprouts from rat aortic rings, in comparison to the unconditioned media. Apparently, the serum level is more relevant than the oxygen level – minor differences were observed in angiogenic properties in conditioned media from MSCs obtained under normal and hypoxic condition (Burlacu et al., 2013).

Tissue-remodeling and anti-fibrotic activity

Scar formation is an inseparable process of wound healing. MSCs have an ability to reduce accumulation of the extracellular matrix proteins. In the secretome from umbilical cord mesenchymal stem cells (UC-MSCs) in mice with fibrosis, 32 proteins were identified which downregulate expression of fibrotic factors, such as the metalloproteinases, collagens, TGF-β and Smad proteins. The milk fat globule EGF factor 8 (MFGE8) negatively regulates expression of TGFβR1 transforming growth factor β type 1 receptor, and therefore acts as an anti-fibrotic factor (An et al., 2017). The hypoxic environment promotes expression of cytoprotective factors in Akt1 overexpressing BM-MSCs (Gnecchi et al., 2005).

MSCs-released vesicles – horizontal material transfer and cell-to-cell communication

Extracellular vesicles (micro vesicles (MVs) and exosomes) are small membrane vesicles (30–150 nm in diameter) of endocytic origin (Simpson et al., 2008). They arise from intracellular membrane of the cell by inward budding and are released from the cells into extracellular space after fusion with the plasma membrane (Mears et al., 2004). During their formation, various transmembrane proteins and cytosolic components are incorporated into membrane invagination and closed within the vesicles (van Niel et al., 2006). It has been found that MSCs can produce higher quantities of exosomes than other cells (Yeo et al., 2013). Extensive composition analysis of EVs released from MSCs has revealed occurrence of over 900 proteins. Examples of some identified proteins are depicted in Table 1. Among the set of cellular proteins, EVs may also contain a variety of lipids and different kinds of RNAs (mRNAs, microRNAs, tRNA, long noncoding RNAs) (http://exocarta.ludwig.edu.au). In general, the exosomes comprise tissue- or cell-type specific molecule composition depending on the tissue type in which they arise. In MSCs-derived EVs, CD73, CD90 and CD105 antigens have been found that are characteristic markers for MSCs (Dominici et al., 2006).

Recently, cell-cell communication mediated by extracellular vesicles has gained scientific interest. It is believed that a possible mechanism of intercellular communication through exosomes follows three pathways (Threy et al., 2001; Janowska-Wieczorek et al., 2005; Bi et al., 2007; Camussi et al., 2010; Raposo & Stahl, 2019):

Several studies have reported that MSCs-derived exosomes perform functions similar to those of MSCs, including repairing tissue and suppressing the inflammatory response. For this reason, MSCs-derived extracellular vesicles seem to be an alternative to MSCs in medical applications. The cell-free therapy offers safety and low immunogenicity with the same regenerative capacities as the mother cells (Rager et al., 2016; Willis et al., 2018). The use of MSC-conditioned media without cell transplantation can promote tissue repair, which supports a potential role of EVs in regenerative medicine (Chen et al., 2014; Tsai et al., 2014).

Fusion – direct cell-cell interaction

One of the possible mechanisms of MSCs action may be fusion with the host cells. Cell fusion is an omnipresent process in life and there is also evidence for fusion processes between stem cells and other neighboring cells.

The mechanism of stem cells fusion with other tissue cells remains elusive as potentially therapeutic. The discussion is still ongoing, partially because of the frequency of fusion events that are very rare. This is probably the main reason why we can find only a few papers describing the fusion process of MSCs. Several studies have reported that stem cells can fuse with cardiomyocytes either by a permanent or partial cell fusion process (Song et al., 2011; Kouris et al., 2012; Shadrin et al., 2015). Shadrin et al. (2015) found that heterologous cell fusion promoted cardiomyocyte reprogramming back to a progenitor-like state, and showed that stem cell mitochondria were transferred into cardiomyocytes and persisted in hybrids. The available data concerns mainly bone marrow derived MSCs (BM-MSCs) fusion. It has been reported that in the injured tissues, BM-MSCs can fuse in vivo with differentiated cells and form hybrids with a regenerative potential (Freeman et al., 2015). The bone marrow-derived hybrids were found in many organs, such as the brain, retina, liver, muscle, and gut, where they participated in the reestablishment of tissue function (Acquistapace et al., 2011; Song et al., 2011; Kouris et al., 2012; Freeman et al., 2015).

Migration and homing – crucial processes in MSCs-based therapy

Overcoming the endothelial barrier

Migration and homing of transplanted cells is a very complicated process that is still under investigation. Depending on the type of tissue source from which the MSCs are isolated, they have their unique, individual ability to migrate and home in a place of damage. These properties of MSCs are crucial for stem cell-based therapies (Pendleton et al., 2013; Chen et al., 2014; Ullah et al., 2015). The directional migration is not a random process but it occurs in response to factors (i.e. chemokines) termed as chemoattractants (Russo et al., 2014). The chemokine gradient released from the injured site recruits transplanted MSCs and provokes them to directly migrate to the target site, which we generally refer to as chemotaxis (Yoon et al., 2016). The mechanism of MSCs migration may vary depending on the method of MSCs administration. Local transplantation in spatial proximity to the injured site causes the cells to circumvent the long migration route through the vascular system. Intravenous (systemic) injection is favored but accompanied by more adverse events (e.g. pulmonary microembolism) (Boltze et al., 2015), and requires overcoming of the endothelial barrier. Many reports describe MSCs migration as highly resembling the leukocyte migration model. According to this scenario, systemic homing is a multistep process involving rolling, crawling, adhesion and extravasation (diapedesis). In the right spot the MSCs integrate with endothelial cells, then overcome the endothelial basement membrane and the pericyte sheath, and then continue the interstitial migration towards the target site, that is navigated by the chemokine gradient (Laird et al., 2008; Teo et al., 2012; Schmidt et al., 2013; Ullah et al., 2015).

Factors involved in MSCs migration

Many chemokines and their receptors located on the cells’ surface are involved in the migration of MSCs and their homing at the target site (Cong et al., 2014). The inflammation and inflammatory processes play an important role in MSCs migration (Guan et al., 2018; Su et al., 2018). The stromal cell-derived factor-1 alpha chemokine (SDF-1α) and its receptor (CXCR4) play a crucial role in the inflammatory process. The SDF-1α protein acts as a chemoattractant that mediates the recruitment of circulating MSCs that express the chemokine type 4 receptor. It can also affect expression of other factors, such as the hepatocyte growth factor (HGF), stem cell factor (SCF) or Fms-related tyrosine kinase 3 ligand, by which cells are targeted to the place of damage (Kitaori et al., 2009; Baek et al., 2011; Wang et al., 2017; Su et al., 2018). Several research groups have shown that growth factors, such as PDGF, TNF-α, and TGF-β1, have a strong chemoattractive effect on MSCs, which enhances their activity for directional migration (Wang et al., 2017). The studies conducted so far have shown that the damaged tissues inside the body secrete numerous cytokines and express factors (chemoattractants) that guide MSCs to the site of damage.

It turns out that not only the source of stem cells and the number of passages, but also the method of cell isolation and culture can have a relevant impact on the migratory potential of MSCs (Ode et al., 2011). It has been shown that an important issue to consider in the migration studies is the appropriate method of culture and medium supplementation, which allows the cells to optimally grow and release factors that are pivotal during the MSCs migration process after their transplantation (Fu et al., 2019). The MMP matrix metalloproteinases that are necessary for efficiency migration and homing of MSCs play an important role in this process. MMPs belong to the proteolytic enzyme family that participates in degradation of the basement membrane and extracellular matrix (ECM) (Ries et al., 2007). The matrix metalloproteinase-1 (MMP1), acting through the MMP1/PAR1 axis, causes decay of the interstitial collagen type I, II and III (Ho et al., 2009; Pan et al., 2014). It has been proven that upregulation of MMP1 expression leads to a high migration capacity of MSCs. In vitro studies have shown that knockdown of the MMP1 gene in MSCs results in inhibition of their migration capacity (Pan et al., 2014). Thus, cells with high MMP1 expression can be a valuable agent in clinical therapy affecting the possibility of cell engraftment at the target site.

During the MSCs in vitro culture, expression of factors involved in the migration process, such as CXCR4, may significantly decrease (Richter et al., 2017). To prevent this effect, the MSCs culture medium can be supplemented with cytokines that stimulate cells and preserve the potential of these cells for migration and homing. It was observed that addition of a cytokine cocktail containing HGF or SCF to the culture medium resulted in improvement of directional migration of cells after administration, as well as homing at the target site (Shi et al., 2007; Richter et al., 2017; Zhang et al., 2017). The oxygen culture conditions, i.e. hypoxia, may lead to an increase in the CXCR4 protein expression, which consequently promotes and activates the migration process (Liu et al., 2010).

The process of stem cell migration depends on the microenvironment in which they are found. Factors secreted during tissue damage lead to the recruitment of the transplanted cells to this site, which is pivotal from the point of view of cellular therapies (Zhang et al., 2017). The CD44 and CXCR4 proteins contribute significantly to this mechanism. The imprisonment of MSCs in the lungs, liver or spleen after intravenous infusion, constitutes a serious obstacle in cellular therapies. For this reason, therapies often use administration of cells directly at the site of injury (Russo et al., 2014). Nevertheless, stem cell properties allow for efficient migration and colonization at the site of injury. Initiation of the migration process can take place not only through mechanical stimuli (e.g. acute shear stress) but also by activation of receptors located at the MSCs surface, even through factors at a large distance from the target site (Kim et al., 2013; Artemenko et al., 2016). Activated MSCs enter the appropriate signaling pathway which initiates the migration and adhesion processes, and adaptation to the ECM environment (Kim et al., 2013; Russo et al., 2014).

Adhesion molecules – key players in migration and homing of mscs

Several classes of cell adhesion molecules mediate interaction between the cell and the substratum. Integrins are the most important among them. They participate in the cell-substratum and cell-cell interactions and are present on almost all human cells (Albelda and Buck, 1990). The MSCs adhesion ability differs due to a different expression of certain integrins, such as α1, α 2, α3, α4, α5, α6, αV, β1, and β2 (Semon et al., 2010; Schmidt et al., 2013). FACS analyses of functional characterization of MSCs described integrins αV, α1, α 2, α3, α5, and α6, but not α4, β, β4, β1, and β2 (Uder et al., 2018). MSCs may feature different adhesion properties depending on the tissue source, isolation and culture procedures (Barry & Murphy, 2004; Lin et al., 2006; Rui et al., 2010; Ren et al., 2015; Sanjurjo-Rodriguez et al., 2017; Uder et al., 2018). Many studies demonstrated that β1 integrins are important for the intramyocardial trafficking of MSCs (Ip et al., 2008), and are crucial for the rolling and adhesion of MSCs (Ruster et al., 2006).

Additionally, a study by Ip and others (Ip et al., 2008) suggested that the very late antigen-4 (VLA-4) and vascular cell adhesion molecule-1 (VCAM-1) are expressed in MSCs (Ip et al., 2008). Among them, both the α4 and β1 subunits play a crucial role in VLA-4 (the integrin very late antigen) formation. Nitzsche and others and Chamberlain and others (Chamberlain et al., 2007; Nitzsche et al., 2017) describe the VLA-4 involvement in the endothelial rolling and apprehension at the inflammation sites. Both authors cite the results of experiments confirming the VCAM-1’s molecule role in firm adhesion of MSCs. In many investigations, blocking the VCAM-1 or VLA-4 mediator, had significantly inhibited adhesion of MSCs to the endothelial cells (D’Ambrosio et al., 1998; Segers et al., 2006). These studies have verified that MSCs bind to endothelial cells in a P-selectin dependent manner. Ruster and others (Ruster’s et al., 2006) showed that rolling of MSCs engages VLA-4/VCAM-1 to provide firm adhesion to the endothelial cells (Ruster et al., 2006).

Moreover, the study of Ruster’s and others (Ruster’s et al., 2006) presents P-selectin involvement in the extravasation cascade – as it has been proven in the MSCs animal model (Ruster et al., 2006). In this study, the P-selectin deficient mice show a decrease in MSCs rolling along the walls of the blood vessels in the ear veins. Similar observations have been also made in the umbilical vein endothelial in vitro model, where MSCs rolling has decreased by neutralizing the P-selectin antibody. The L-selectin expression is described as insignificant in the case of MSCs rolling on endothelium during the first stage of the recruitment process. In comparison to leukocytes, their expression on MSCs is much lower. Additionally, no expression of PECAM-1/CD31 (platelet/endothelial cell adhesion molecule 1) has been found on MSCs (Bruder et al., 1998; Pittenger et al., 1999; Shur et al., 2002; Ruster et al., 2006), whereas occurrence of hematopoietic cell L-/E-selectin ligand, as well as NCAM1/CD56 (neural adhesion molecule 1), ICAM2/CD102 (intercellular adhesion molecule 2), MCAM/CD146 (melanoma cell adhesion molecule 2), and ALCAM/CD166 (activated leukocyte cell adhesion molecule), and ICAM1/CD54 or ICAM3/CD50 has been shown. In addition, the P-selectin antigens are not present on the surface of MSCs (Uder et al., 2018). In contrast to the Minguell and others (Minguell et al., 2001) results, Krampera and others (Krampera et al., 2006) described expression not only of VCAM-1 and ALCAM, but also of ICAM-1 and ICAM-3, and in addition that of endoglin/CD105 (Minguell et al., 2001; Krampera et al., 2006). As it has been previously mentioned, the existing differences in expression may be the consequence of experimental conditions, e.g. cell culture or their source. Segers et al. (2006) described a significant role of VCAM-1 in the animal rat model where they have stimulated the cardiac microvascular endothelial cells and then they have monitored adhesion of MSCs to them (Segers et al., 2006). There is no proven role for the ICAM adhesion molecule in this model.

Molecular mechanism of MSCs adhesion

Hypoxia, acute burns and injury of skeletal muscle, all trigger mobilization of MSCs into peripheral blood (Mansilla et al., 2006; Ramirez et al., 2006; Rochefort et al., 2006). Differences in the cell adhesion molecule pattern, under different culture conditions, may be a starting point for a discussion about the MSCs homing mechanism. At present, there is no detailed and strong data describing MSCs transendothelial migration. Because of existing differences in expression of the cell adhesion molecules between MSCs and well-described leukocytes, there is a need for further detailed investigation of MSCs adhesion, rolling, diapedesis and homing.

Regarding the mechanism of the homing process, active surface molecules that are involved in MSCs – endothelium adhesion are in the center of interest. They are believed to be involved in the principal mechanism of this process (Karp & Leng, 2009). As expression of certain chemokine receptors during the extravasation process is widely described, it is still unclear what their exact involvement in this process is.

The presence of CCR2, CCR4, CCR7, CCR10, CXCR5, CXCR6, and CXCR4 is referred to in the literature (Wynn et al., 2004; Ringe et al., 2007; Andreas et al., 2014; Nitzsche et al., 2017). CCR2 participates in organ-specific homing (Belema-Bedada et al., 2008; Guo et al., 2013). In the Smith’s and others (Smith’s et al., 2012) research, chemokines: CXCL12, CXCL13, CXCL16, CCL11, and CCL22 are described as the ligands for the chemokine receptors: CXCR4, CXCR5, CXCR6, CCR3, and CCR4, which have been identified on the MSCs. In this study, the authors show a significantly enhanced transendothelial migration across the bone marrow endothelial cells (Smith et al., 2012). On the other hand, in the research by Ip and others (Ip et al., 2007), CXCR4 is not directly involved in the MSCs transendothelial migration, despite its proven expression on these cells. One of the reasons for such inconsistency in the role of specific cytokines may be rooted in the previously mentioned differences in experimental conditions that vary between the studies, including cell culturing, type of medium, use of serum etc., as well as the isolation procedures (Uder et al., 2018).

Two signaling pathways are established to be of a great importance for the MSCs transendothelial migration. The phosphoinositide 3-kinase (PI3K)/Act pathway and phosphokinase C (PKC) pathway are described to have a significant role in transendothelial migration. Among the different kinases involved in the MSCs signaling, the integrin-linked kinase (ILK) is indicated. Its role as an intracellular adapter in transmission of signals from the outside to the inside is discussed in the literature (Fig. 2) (Schmidt et al., 2006; Picinich et al., 2010; Widmaier et al., 2012).

The influence of oxygen concentration on biological properties of MSCs

The important issue in the investigation of biological properties of MSC is the microenvironment (niche) in which the cells reside. One of the crucial factors is oxygen concentration. It should be noted that the standard culture conditions in vitro including 21% O2 (21 kPa/160 mmHg) are not identical to the in vivo environment, where oxygen concentration is significantly lower, about 2–9%, depending on the vascularization of the tissue and its direct contact with air (Brahimi-Horn & Pouyssegur, 2007; Ward, 2008). In the bone marrow, as a primary source of stem cells (both hematopoietic and mesenchymal), the concentration levels of O2 have been reported to range from 1.5% to 7% (Spencer et al., 2014). A change in the oxygen level in an in vitro culture leads to alterations in metabolism (secretion of signaling factors – growth factors and cytokines), proliferation (self-renewal), motility (adhesion, migration), and differentiation potential. Consequently, the stemness potential can be lost.

A key factor that plays a crucial role in the regulation of stem cells metabolism is HIF-1α (hypoxia-inducible factor 1 alpha), a transcription factor involved in cellular response to low oxygen availability. HIF-1α is expressed by cells in a hypoxia environment and makes their metabolism pathways similar to those occurring in an in vivo niche (Takubo et al., 2010). These facts explain why the oxygen level should be particularly taken into account during cell expansion for clinical applications.

The results of performed studies have revealed that hypoxia or HIF-1α stabilization have a beneficial effect on MSCs functions, such as proliferation, migration and adhesion, regardless of the tissue type of their origin. Low oxygen concentration favors maintenance of MSCs physiological properties, including proper expression of surface receptors involved in MSCs migration and adhesion (Li et al., 2013; Choi et al., 2016). Song et al. (2009) presented that hypoxia promotes MSCs adhesion to myocardium, thereby increasing their therapeutic potential (Song et al., 2009).

Many studies have also observed a hypoxia dependent effect on the paracrine activity of MSCs. For example, secretion of the vascular endothelial growth factor (VEGF) by AD-MSCs (adipose tissue derived-MSCs) is considerably enhanced under hypoxic conditions, where HIF-1α is more stable (Kang et al., 2014). VEGF, apart from its proangiogenic properties, also stimulates motility of stem cells (Yun et al., 2009).

There is also evidence that the proliferation capacity of MSCs is significantly improved in an environment with low oxygen concentration (Lech et al., 2016; Choi et al., 2017). Some studies suggest that a low oxygen level promotes self-renewal (asymmetric) divisions and inhibits symmetric ones, which precedes cell differentiation (Liu et al., 2012). The reduced oxygen content during stem cell culture has also a beneficial effect on their genomic stability. Lech and others (Lech at al., 2016) have showed that AD-MSCs cultured in reduced oxygen content (5% O2) maintain a correct karyotype profile compared to cells cultured in normoxia condition. In cultures growing under 21% O2 concentration numerous karyotyping abnormalities (i.e. chromosome polyploidy and haploid chromosomes) have been observed (Lech et al., 2016).

As detailed above, hypoxia promotes kinetic growth, genetic stability, proper pattern of MSC-specific surface markers, and increases cell lifespan, as well as exerts a positive effect on the paracrine activity and adhesion properties. Thus, conducting cell culture at the physiological oxygen level seems to be more effective and safe in the case of MSCs used in regenerative therapies.

Conclusions

Mesenchymal stem cells are undoubtedly the future of regenerative medicine. The benefits of their clinical use have been proven in many studies, but development of safe and efficient therapy requires future investigations. The therapeutic potential is a result of their unique properties, including the paracrine activity, directional migration, adhesion and homing at the site of injury. A particular advantage of MSCs when compared to pharmaceutical agents is their capability to secrete a cocktail of bioactive factors in response to circumstances and microenvironmental conditions. It should be noted that the culture conditions and the cell source have fundamental influence on biological properties of MSCs which should be considered in clinical use of MSCs.

Conflicts of Interest

The authors declare no conflict of interest.

References

Acquistapace A, Bru T, Lesault PF, Figeac F, Coudert AE, le Coz O, Christov C, Baudin X, Auber F, Yiou R, Dubois-Randé JL, Rodriguez AM (2011) Human mesenchymal stem cells reprogram adult cardiomyocytes toward a progenitor-like state through partial cell fusion and mitochondria transfer. Stem Cells 29: 812–824. https://doi.org/10.1002/stem.632.

Albelda S, Buck C (1990) Integrins and other cell adhesion molecules. FASEB Fed Am Soc Exp Biol J 4: 2868–2880. PMID: 2199285

An SY, Jang YJ, Lim HJ, Han J, Lee J, Lee G, Park JY, Park SY, Kim JH, Do BR, Han C, Park HK, Kim OH, Song MJ, Kim SJ, Kim JH (2017) Milk fat globule-EGF factor 8, secreted by mesenchymal stem cells, protects against liver fibrosis in mice. Gastroenterology 152: 1174–1186. https://doi.org/10.1053/j.gastro.2016.12.003

Andreas K, Sittinger M, Ringe J (2014) Toward in situ tissue engineering: Chemokine-guided stem cell recruitment. Trends Biotechnol 32: 483–492. https://doi.org/10.1016/j.tibtech.2014.06.008

Artemenko Y, Axiotakis L, Borleis J, Iglesias PA, Devreotes PN (2016) Chemical and mechanical stimuli act on common signal transduction and cytoskeletal networks. Proc Natl Acad Sci 113: e7500–e7509. https://doi.org/10.1073/pnas.1608767113.

Baek SJ, Kang SK, Ra JC (2011) In vitro migration capacity of human adipose tissue-derived mesenchymal stem cells reflects their expression of receptors for chemokines and growth factors. Exp Mol Med 43: 596–603. https://doi.org/10.3858/emm.2011.43.10.069.

Barry FP, Murphy JM (2004) Mesenchymal stem cells: Clinical applications and biological characterization. Int J Biochem Cell Biol 36: 568–584. https://doi.org/10.1016/j.biocel.2003.11.001

Belema-Bedada F, Uchida S, Martire A, Kostin S, Braun T (2008) Efficient homing of multipotent adult mesenchymal stem cells depends on FROUNT-mediated clustering of CCR2. Cell Stem Cell 2: 566–575. https://doi.org/10.1016/j.stem.2008.03.003

Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG (2007) Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol 18: 2486–2496. https://doi.org/10.1681/ASN.2007020140

Boltze J, Arnold A, Walczak P, Jolkkonen J, Cui L, Wagner DC (2015) The dark side of the force – constraints and complications of cell therapies for stroke. Front Neurol 20: 155–171. https://doi.org/10.3389/fneur.2015.00155

Brahimi-Horn MC, Pouyssegur J (2007) Oxygen, a source of life and stress. FEBS Lett 581: 3582–3591. https://doi.org/10.1016/j.febslet.2007.06.018

Brini AT, Amodea G, Ferreira LM, Milani A, Niada S, Moschetti G, Franchi S, Borsani E, Rodella LF, Panerai AE, Sacerdote P (2017) Therapeutic effect of human adipose-derived stem cells and their secretome in experimental diabetic pain. Sci Rep 7: 1–15. https://doi.org/10.1038/s41598-017-09487-5

Bruder SP1, Ricalton NS, Boynton RE, Connolly TJ, Jaiswal N, Zaia J, Barry FP (1998) Mesenchymal stem cell surface antigen SB-10 corresponds to activated leukocyte cell adhesion molecule and is involved in osteogenic differentiation. J Bone Miner Res 13: 655–663. https://doi.org/10.1359/jbmr.1998.13.4.655

Burlacu A, Grigorescu G, Rosca AM, Preda MB, Simionescu M (2013) Factors secreted by mesenchymal stem cells and endothelial progenitor cells have complementary effects on angiogenesis in vitro. Stem Cells and Dev 22: 643–653. https://doi.org/10.1089/scd.2012.0273

Camussi G, Deregibus MC, Bruno S, Cantaluppi V, Biancone L (2010) Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int 78: 838–848. https://doi.org/10.1038/ki.2010.278

Chamberlain G, Fox J, Ashton B, Middleton J (2007) Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 25: 2739–2749. https://doi.org/10.1634/stemcells.2007-0197

Chen G, Yue A, Ruan Z, Yin Y, Wang R, Ren Y, Zhu L (2014) Monitoring the biology stability of human umbilical cord-derived mesenchymal stem cells during long-term culture in serum-free medium. Cell Tissue Bank 15: 513–521. https://doi.org/10.1007/s10561-014-9420-6

Chen L, Xu Y, Zhao J, Zhang Z, Yang R, Xie J, Liu X, Qi S (2014) Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS One 9: e96161. https://doi.org/10.1371/journal.pone.0096161

Choi W, Kwon SJ, Jin HJ, Jeong SY, Choi SJ, Oh W, Yang YS, Jeon HB, Jeon ES (2017) Optimization of culture conditions for rapid clinical-scale expansion of human umbilical cord blood-derived mesenchymal stem cells. Clin Transl Med 6: 38. https://doi.org/10.1186/s40169-017-0168-z

Choi JH, Lim SM, Yoo YI, Jung J, Park JW, Kim GJ (2016) Microenvironmental interaction between hypoxia and endothelial cells controls the migration ability of placenta-derived mesenchymal stem cells via alpha4 integrin and rho signaling. J Cell Biochem 117: 1145–1157. https://doi.org/10.1002/jcb.25398

Cong Q, Yeh J, Xia XC, Mishina Y, Hao AJ, Li BJ (2014) PDGF-AA promotes osteogenic differentiation and migration of mesenchymal stem cell by down-regulating PDGFRa and derepressing BMP-Smad1 signaling. PLoS One 9: 113–128. https://doi.org/10.1371/journal.pone.0113785

Crigler L, Robey RC, Asawachaicharn A, Gaupp D, Phinney DG (2006) Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol 198: 54–64

D’Ambrosio D1, Iellem A, Bonecchi R, Mazzeo D, Sozzani S, Mantovani A, Sinigaglia F (1998) Selective up-regulation of chemokine receptors CCR4 and CCR8 upon activation of polarized human type 2 Th cells. J Immunol 161: 5111–5115

Meirelles LdaS, Fontes AM, Covas DT, Caplan AI (2009) Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev 20: 419–427. https://doi.org/10.1016/j.cytogfr.2009.10.002

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS, Deans RJ, Keating A, Prockop DJ, Horwitz EM (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315–317. https://doi.org/10.1080/14653240600855905

Doorn J, Moll G, Le Blanc K, van Blitterswijk C, de Boer J (2012) Therapeutic applications of mesenchymal stromal cells: paracrine effects and potential improvements. Tissue Eng Part B Rev 18: 101–115. https://doi.org/10.1089/ten.TEB.2011.0488

Fei X, Jiang S, Zhang S, Li Y, Ge J, He B, Goldstein S, Ruiz G (2013) Isolation, culture, and identification of amniotic fluid-derived mesenchymal stem cells. Cell Biochem Biophys 67: 689–694. https://doi.org/10.1007/s12013-013-9558-z

Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Gonçalves RM (2018) Mesenchymal stromal cell secretome: Influencing therapeutic potential by cellular pre-conditioning. Front Immunol 9: 2837. https://doi.org/10.3389/fimmu.2018.02837

Freeman BT, Kouris NA, Ogle BM (2015) Tracking fusion of human mesenchymal stem cells after transplantation to the heart. Stem Cells Transl Med 4: 685–694. https://doi.org/10.5966/sctm.2014-0198

Fu X, Liu G, Halim A, Ju Y, Luo Q, Song G (2019) Mesenchymal stem cell migration and tissue repair. Cells 8: 784–799. https://doi.org/10.3390/cells8080784.

Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, Noiseux N, Zhang L, Pratt RE, Ingwall JS, Dzau VJ (2005) Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells Nat Med 11: 367–368. https://doi.org/10.1038/nm0405-367

Guan SP, Lam AT, Newman JP, Chua KL, Kok CY, Chong ST, Chua MK, Lam PY (2018) Matrix metalloproteinase-1 facilitates MSC migration via cleavage of IGF-2/IGFBP2 complex. FEBS Open Bio 8: 15–26. https://doi.org/10.1002/2211-5463.12330

Guo J, Zhang H, Xiao J, Wu J, Ye Y, Li Z, Zou Y, Li X (2013) Monocyte chemotactic protein-1 promotes the myocardial homing of mesenchymal stem cells in dilated cardiomyopathy. Int J Mol Sci 14: 8164–8178. https://doi.org/10.3390/ijms14048164

Hare JM, DiFede DL, Rieger AC, Florea V, Landin AM, El-Khorazaty J, Khan A, Mushtaq M, Lowery MH, Byrnes JJ, Hendel RC, Cohen MG, Alfonso CE, Valasaki K, Pujol MV, Golpanian S, Ghersin E, Fishman JE, Pattany P, Gomes SA, Delgado C, Miki R, Abuzeid F, Vidro-Casiano M, Premer C, Medina A, Porras V, Hatzistergos KE, Anderson E, Mendizabal A, Mitrani R, Heldman AW (2017) Randomized comparison of allogeneic versus autologous mesenchymal stem cells for nonischemic dilated cardiomyopathy: POSEIDON-DCM trial. J Am Coll Cardiol 69: 526–537. https://doi.org/10.1016/j.jacc.2016.11.009

Heidari B, Shirazi A, Akhondi MM, Hassanpour H, Behzadi B, Naderi MM, Sarvari A, Borjian S (2013) Comparison of proliferative and multilineage differentiation potential of sheep mesenchymal stem cells derived from bone marrow, liver, and adipose tissue. Avicenna J Med Biotechnol 5: 104–117. PMCID: PMC3689554

Ho IA, Chan KY, Ng WH, Guo CM, Hui KM, Cheang P, Lam PY (2009) Matrix metalloproteinase 1 is necessary for the migration of human bone marrow-derived mesenchymal stem cells toward human glioma. Stem Cells 27: 1366–1375. https://doi.org/10.1002/stem.50.

Ip JE, Wu Y, Huang J, Zhang L, Pratt RE, Dzau VJ (2009) Mesenchymal stem cells use integrin beta1 not CXC chemokine receptor 4 for myocardial migration and engraftment. Mol Biol Cell 18: 2873–2882. https://doi.org/10.1091/mbc.e07-02-0166

Janowska-Wieczorek A,Wysoczynski M, Kijowski J, Marquez-Curtis L, Machalinski B, Ratajczak J, Ratajczak MZ (2005) Microvesicles derived from activated platelets induce metastasis and angiogenesis inlung cancer. Int J Cancer 113: 752–760. https://doi.org/10.1002/ijc.20657

Karp JM, Leng Teo GS (2009) Mesenchymal stem cell homing: The devil is in the details. Cell Stem Cell 4: 206–216. https://doi.org/10.1016/j.stem.2009.02.001

Kim YS, Noh MY, Kim JY, Yu HJ, Kim KS, Kim SH, Koh SH (2013) Direct GSK-3β inhibition enhances mesenchymal stromal cell migration by increasing expression of beta-PIX and CXCR4. Mol Neurobiol 47: 811–820. https://doi.org/10.1007/s12035-012-8393-3

Kitaori T, Ito H, Schwarz EA, Tsutsumi R, Yoshitomi H, Oishi S, Nakano M, Fujii N, Nagasawa T, Nakamura T (2009) Stromal cell-derived factor 1/CXCR4 signaling is critical for the recruitment of mesenchymal stem cells to the fracture site during skeletal repair in a mouse model. Arthritis Rheumatol 60: 813–823. https://doi.org/10.1002/art.24330

Kouris NA, Schaefer JA, Hatta M, Freeman BT, Kamp TJ, Kawaoka Y, Ogle BM (2012) Directed fusion of mesenchymal stem cells with cardiomyocytes via VSV-G facilitates stem cell programming. Stem Cells Int 2012: 414038. https://doi.org/10.1155/2012/414038

Krampera M, Pizzolo G, Aprili G, Franchini M (2006) Mesenchymal stem cells for bone, cartilage, tendon and skeletal muscle repair. Bone 39: 678–683. https://doi.org/10.1016/j.bone.2006.04.020

Laird DJ, von Andrian UH, Wagers AJ (2008) Stem cell trafficking in tissue development, growth, and disease. Cell 132: 612–630. https://doi.org/10.1016/j.cell.2008.01.041

Lech W, Figiel-Dabrowska A, Sarnowska A, Drela K, Obtulowicz P, Noszczyk BH, Buzanska L, Domanska-Janik K (2016) Phenotypic, functional, and safety control at preimplantation phase of MSC-based therapy. Stem Cell Int 2016: 2514917. https://doi.org/10.1155/2016/2514917.

Lee JK, Jin HK, Endo S, Schuchman EH, Carter JE, Bae JS (2010) Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduces amyloid-beta deposition and rescues memory deficits in Alzheimer’s disease mice by modulation of immune responses. Stem Cells 28: 329–343. https://doi.org/10.1002/stem.277

Li Q, Wang Y, Deng Z (2013) Pre-conditioned mesenchymal stem cells: A better way for cell-based therapy. Stem Cell Res Ther 4: 63. https://doi.org/10.1186/scrt213

Lin Y, Liu L, Li Z, Qiao J, Wu L, Tang W, Zheng X, Chen X, Yan Z, Tian W (2006) Pluripotency potential of human adipose-derived stem cells marked with exogenous green fluorescent protein. Mol Cell Biochem 291: 1–10. https://doi.org/10.1007/s11010-006-9188-5

Liu H, Xue W, Ge G, Luo X, Li Y, Xiang H, Ding X, Tian P, Tian X (2010) Hypoxic preconditioning advances CXCR4 and CXCR7 expression by activating HIF-1α in MSCs. Biochem Biophys Res Commun 401: 509–515. https://doi.org/10.1016/j.bbrc.2010.09.076

Liu W, Wen Y, Bi P, Lai X, Liu XS, Liu X, Kuang S (2012) Hypoxia promotes satellite cell self-renewal and enhances the efficiency of myoblast transplantation. Development 139: 2857–2865. https://doi.org/10.1242/dev.079665

Mansilla E, Marin GH, Drago H, Sturla F, Salas E, Gardiner C, Bossi S, Lamonega R, Guzmán A, Nuñez A, Gil MA, Piccinelli G, Ibar R, Soratti C (2006) Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc 38: 967–969. https://doi.org/10.1016/j.transproceed.2006.02.053

Martins LF, Costa RO, Pedro JR, Aguiar P, Serra SC, Teixeira FG, Sousa N, Salgado AJ, Almeida RD (2017) Mesenchymal stem cells secretome-induced axonal outgrowth is mediated by BDNF. Sci Rep 7: 1–13. https://doi.org/10.1038/s41598-017-03592-1

Mears R, Craven RA, Hanrahan S, Totty N, Upton C, Young SL, Patel P, Selby PJ, Banks RE (2004) Proteomic analysis of melanomaderived exosomes by two-dimensional polyacrylamide gel electrophoresis and mass spectrometry. Proteomics 4: 4019–4031. https://doi.org/10.1002/pmic.200400876

Meier RP, Müller YD, Morel P, Gonelle-Gispert C, Bühler LH (2013) Transplantation of mesenchymal stem cells for the treatment of liver diseases, is there enough evidence? Stem Cell Res 11: 1348–1364. https://doi.org/10.1016/j.scr.2013.08.011

Minguell JJ, Erices A, Conget P (2001) Mesenchymal stem cells. Exp Biol Med (Maywood) 226: 507–520. https://doi.org/10.1177/153537020122600603

Mrozik KM, Zilm PS, Bagley CJ, Hack S, Hoffmann P, Gronthos S, Bartold PM (2010) Proteomic characterization of mesenchymal stem cell-like populations derived from ovine periodontal ligament, dental pulp, and bone marrow: analysis of differentially expressed proteins. Stem Cells 19: 1485–1499. https://doi.org/10.1089/scd.2009.0446

Musialek P, Mazurek A, Jarocha D, Tekieli L, Szot W, Kostkiewicz M, Banys RP, Urbanczyk M, Kadzielski A, Trystula M, Kijowski J, Zmudka K, Podolec P, Majka M (2015) Myocardial regeneration strategy using Wharton’s jelly mesenchymal stem cells as an off-the-shelf ‘unlimited’ therapeutic agent: results from the Acute Myocardial Infarction First-in-Man Study. Post Kardiol Interw 11: 100–107. https://doi.org/10.5114/pwki.2015.52282

Musiał-Wysocka A, Kot M, Sułkowski M, Badyra B, Majka M (2019a) Molecular and functional verification of Wharton’s Jelly mesenchymal stem cells (WJ-MSCs) pluripotency. Int J Mol Sci 20: 1807. https://doi.org/10.3390/ijms20081807

Musiał-Wysocka A, Kot M, Sułkowski M, Majka M (2019b) Regenerative potential of the product “CardioCell” derived from the Wharton’s jelly mesenchymal stem cells for treating hindlimb ischemia. Int J Mol Sci 20: 4632. https://doi.org/10.3390/ijms20184632

Najar M, Raicevic G, Boufker HI, Fayyad Kazan H, De Bruyn C, Meuleman N, Bron D, Toungouz M, Lagneaux L (2010) Mesenchymal stromal cells use PGE2 to modulate activation and proliferation of lymphocyte subsets: Combined comparison of adipose tissue, Wharton’s Jelly and bone marrow sources. Cell Immunol 264: 171–179. https://doi.org/10.1016/j.cellimm.2010.06.006

Nitzsche F, Müller C, Lukomska B et al. (2017) Concise review: MSC adhesion cascade-insights into homing and transendothelial migration. Stem Cells 35: 1446–1460. https://doi.org/10.1002/stem.2614

Ode A, Kopf J, Kurtz A, Schmidt-Bleek K, Schrade P, Kolar P, Buttgereit F, Lehmann K, Hutmacher DW, Duda GN, Kasper G (2011) CD73 and CD29 concurrently mediate the mechanically induced decrease of migratory capacity of mesenchymal stromal cells. Eur Cell Mater 22: 26–42. https://doi.org/10.22203/eCM.v022a03

Oskowitz A, McFerrin H, Gutschow M, Carter ML, Pochampally R (2011) Serum-deprived human multipotent mesenchymal stromal cells (MSCs) are highly angiogenic. Stem Cell Res 6: 215–225. https://doi.org/10.1016/j.scr.2011.01.004

Pan H, Vojnits K, Liu TT, Meng F, Yang L, Wang Y, Huard J, Cox CS, Lally KP, Li Y (2014) MMP1 gene expression enhances myoblast migration and engraftment following implanting into mdx/SCID mice. Cell Adh Migr 9: 283–292. https://doi.org/10.4161/19336918.2014.983799

Pendleton C, Li Q, Chesler DA, Yuan K, Guerrero-Cazares H, Quinones-Hinojosa A (2013) Mesenchymal stem cells derived from adipose tissue vs bone marrow: in vitro comparison of their tropism towards gliomas. PLoS One 8: 581–598. https://doi.org/10.1371/journal.pone.0058198

Picinich SC, Glod JW, Banerjee D (2010) Protein kinase C zeta regulates interleukin-8-mediated stromal-derived factor-1 expression and migration of human mesenchymal stromal cells. Exp Cell Res 316: 593–602. https://doi.org/10.1016/j.yexcr.2009.11.011

Praveen KL, Kandoi S, Misra R, S V, K R, Verma RS (2019) The mesenchymal stem cell secretome: A new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine Growth Factor Rev 46: 1–9. https://doi.org/10.1016/j.cytogfr.2019.04.002

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR (1999) Multilineage potential of adult human mesenchymal stem cells. Science 284: 143–147. https://doi.org/10.1126/science.284.5411.143

Rager TM, Olson JK, Zhou Y, Wang Y, Besner GE (2016) Exosomes secreted from bone marrow-derived mesenchymal stem cells protect the intestines from experimental necrotizing enterocolitis. J Pediatr Surg 51: 942–947. https://doi.org/10.1016/j.jpedsurg.2016.02.061

Ramirez M, Lucia A, Gomez-Gallego F, Esteve-Lanao J, Pérez-Martínez A, Foster C, Andreu AL, Martin MA, Madero L, Arenas J, García-Castro J (2006) Mobilisation of mesenchymal cells into blood in response to skeletal muscle injury. Br J Sports Med 40: 719–722. https://doi.org/10.1136/bjsm.2006.028639

Raposo G, Stahl PD (2019) Extracellular vesicles: a new communication paradigm? Nat Rev Mol Cell Biol 20: 509–510. https://doi.org/10.1038/s41580-019-0158-7

Ren J, Wang H, Tran K, Civini S, Jin P, Castiello L, Feng J, Kuznetsov SA, Robey PG, Sabatino M, Stroncek DF (2015) Human bone marrow stromal cell confluence: Effects on cell characteristics and methods of assessment. Cytotherapy 17: 897–911. https://doi.org/10.1016/j.jcyt.2015.03.607

Richter R, Forssmann W, Henschler R (2017) Current developments in mobilization of hematopoietic stem and progenitor cells and their interaction with niches in bone marrow. transfus. Med Hemother 44: 151–164. https://doi.org/10.1159/000477262

Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P (2007) MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood 109: 4055–4063. https://doi.org/10.1182/blood-2006-10-051060.

Ringe J, Strassburg S, Neumann K, Endres M, Notter M, Burmester GR, Kaps C, Sittinger M (2007) Towards in situ tissue repair: Human mesenchymal stem cells express chemokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation with CXCL8 but not CCL2. J Cell Biochem 101: 135–146. https://doi.org/10.1002/jcb.21172

Rochefort GY, Delorme B, Lopez A, Hérault O, Bonnet P, Charbord P, Eder V, Domenech J (2006) Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells 24: 2202–2208. https://doi.org/10.1634/stemcells.2006-0164

Rui YF, Lui PP, Li G, Fu SC, Lee YW, Chan KM (2010) Isolation and characterization of multipotent rat tendon-derived stem cells. Tissue Eng A 16: 1549–1558. https://doi.org/10.1089/ten.TEA.2009.0529

Russo V, Young S, Hamilton A, Amsden BG, Flynn LE (2014) Mesenchymal stem cell delivery strategies to promote cardiac regeneration following ischemic injury. Biomaterials 35: 3956–3974. https://doi.org/10.1016/j.biomaterials.2014.01.075

Ruster B, Gottig S, Ludwig RJ, Bistrian R, Müller S, Seifried E, Gille J, Henschler R (2006) Mesenchymal stem cells (MSCs) display coordinated rolling and adhesion behavior on endothelial cells. Blood 108: 3938–3944. https://doi.org/10.1182/blood-2006-05-025098

Sackstein R (2012) Glycoengineering of HCELL, the human bone marrow homing receptor: Sweetly programming cell migration. Ann Biomed Eng 40: 766–776. https://doi.org/10.1007/s10439-011-0461-8

Sanjurjo-Rodriguez C, Castro-Vinuelas R, Hermida-Gomez T, Fernandez-Vazquez T, Fuentes-Boquete IM, de Toro-Santos FJ, Diaz-Prado SM, Blanco-Garcia FJ (2017) Ovine mesenchymal stromal cells: Morphologic, phenotypic and functional characterization for osteochondral tissue engineering. PLoS One 12: e0171231. https://doi.org/10.1371/journal.pone.0171231

Schmidt A, Ladage D, Steingen C, Brixius K, Schinköthe T, Klinz FJ, Schwinger RH, Mehlhorn U, Bloch W (2006) Mesenchymal stem cells transmigrate over the endothelial barrier. Eur J Cell Biol 85: 1179–1188. https://doi.org/10.1016/j.ejcb.2006.05.015

Schmidt S, Moser M, Sperandio M (2013) The molecular basis of leukocyte recruitment and its deficiencies. Mol Immunol 55: 49–58. https://doi.org/10.1016/j.molimm.2012.11.006

Segers VF, Van Riet I, Andries LJ, Lemmens K, Demolder MJ, De Becker AJ, Kockx MM, De Keulenaer GW (2006) Mesenchymal stem cell adhesion to cardiac microvascular endothelium: Activators and mechanisms. Am J Physiol Heart Circ Physiol 290: H1370–H1377. https://doi.org/10.1152/ajpheart.00523.2005

Semon JA, Nagy LH, Llamas CB et al. (2010) Integrin expression and integrin-mediated adhesion in vitro of human multipotent stromal cells (MSCs) to endothelial cells from various blood vessels. Cell Tissue Res 341: 147–158. https://doi.org/10.1007/s00441-010-0994-4

Shadrin IY, Yoon W, Li L, Shepherd N, Bursac N (2015) Rapid fusion between mesenchymal stem cells and cardiomyocytes yields electrically active, non-contractile hybrid cells. Sci Rep 5: 12043. https://doi.org/10.1038/srep12043

Shi M, Li J, Liao L, Chen B, Li B, Chen L, Jia H, Zhao RC (2007) Regulation of CXCR4 expression in human mesenchymal stem cells by cytokine treatment: Role in homing efficiency in NOD/SCID mice. Haematologica 92: 897–904. https://doi.org/10.3324/haematol.10669

Shur I, Marom R, Lokiec F, Socher R, Benayahu D (2002) Identification of cultured progenitor cells from human marrow stroma. J Cell Biochem 87: 51–57. https://doi.org/10.1002/jcb.10267

Simpson RJ, Jensen SS, Lim JW (2008) Proteomic profiling of exosomes: current perspectives. Proteomics 8: 4083–4099. https://doi.org/10.1002/pmic.200800109

Smith H, Whittal C, Weksler B, Middleton J (2012) Chemokines stimulate bidirec-tional migration of human mesenchymal stem cells across bone mar-row endothelial cells. Stem Cells Dev 21: 476–486. https://doi.org/10.1089/scd.2011.0025

Spencer JA, Ferraro F, Roussakis E, Klein A, Wu J, Runnels JM, Zaher W, Mortensen LJ, Alt C, Turcotte R, Yusuf R, Côté D, Vinogradov SA, Scadden DT, Lin CP (2014) Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature 508: 269–273. https://doi.org/10.1038/nature13034

Song SW, Chang W, Song BW, Song H, Lim S, Kim HJ, Cha MJ, Choi E, Im SH, Chang BC, Chung N, Jang Y, Hwang KC (2009) Integrin-linked kinase is required in hypoxic mesenchymal stem cells for strengthening cell adhesion to ischemic myocardium. Stem Cells 27: 1358–1365. https://doi.org/10.1002/stem.47

Song YH, Pinkernell K, Alt E (2011) Stem cell induced cardiac regeneration: fusion/mitochondria exchange and/or transdifferetiation? Cell Cycle 10: 2281–2286. https://doi.org/10.4161/cc.10.14.16513

Su P, Tian Y, Yang C, Ma X, Wang X, Pei J, Qian A (2018) Mesenchymal stem cell migration during bone formation and bone diseases therapy. Int J Mol Sci 19: 2343–2357. https://doi.org/10.3390/ijms19082343.

Takubo K, Goda N, Yamada W, Iriuchishima H, Ikeda E, Kubota Y, Shima H, Johnson RS, Hirao A, Suematsu M, Suda T (2010) Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. Cell Stem Cell 7: 391–402. https://doi.org/10.1016/j.stem.2010.06.020

Teixeira FG, Carvalho MM, Sousa N, Salgado AJ (2013) Mesenchymal stem cells secretome: A new paradigm for central nervous system regeneration? Cell Mol Life Sci 70: 3871–3882. https://doi.org/10.1007/s00018-013-1290-8

Teo GSL, Ankrum JA, Martinelli R et al. (2012) Mesenchymal stem cells transmigrate between and directly through tumor necrosis factor-alpha-activated endothelial cells via both leukocyte-like and novel mechanisms. Stem Cells 30: 2472–2486. https://doi.org/10.1002/stem.1198

Thankamony SP, Sackstein R (2011) Enforced hematopoietic cell E- and L-selectin ligand (HCELL) expression primes transendothelial migration of human mesenchymal stem cells. Proc Natl Acad Sci U S A 108: 2258–2263. https://doi.org/10.1073/pnas.1018064108

Thery C, Boussac M, Veron P, Ricciardi-Castagnoli P, Raposo G, Garin J, Amigorena S (2001) Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol 166: 7309–7318. https://doi.org/10.4049/jimmunol.166.12.7309

Tsai MJ, Tsai SK, Hu BR, Liou DY, Huang SL, Huang MC, Huang WC, Cheng H, Huang SS (2014) Recovery of neurological function of ischemic stroke by application of conditioned medium of bone marrow mesenchymal stem cells derived from normal and cerebral ischemia rats. J Biomed Sci 21: 5. https://doi.org/10.1186/1423-0127-21-5

Uder C, Bruckner S, Winkler S, Tautenhahn HM, Christ B (2018) Mammalian MSC from selected species: Features and applications. Cytometry A 93: 32–49. https://doi.org/10.1002/cyto.a.23239

Ullah I, Subbarao RB, Rho GJ (2015) Human mesenchymal stem cells – current trends and future prospective. Biosci Rep 35: 191–205. https://doi.org/10.1042/BSR20150025.

van Niel G, Porto-Carreiro I, Simoes S, Raposo G (2006) Exosomes: a common pathway for a specialized function. J Biochem (Tokyo) 140: 13–21. https://doi.org/10.1093/jb/mvj128

Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R (2017) Mesenchymal stem cell secretome: Toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci 18: 1852. https://doi.org/10.3390/ijms18091852

Wang S, Qu X, Zhao RC (2012) Clinical applications of mesenchymal stem cells. J Hematol Oncol 5: 19. 10.1186/1756-8722-5-19

Wang Y, Xu J, Zhang X, Wang C, Huang Y, Dai K, Zhang X (2017) TNF-α-induced LRG1 promotes angiogenesis and mesenchymal stem cell migration in the subchondral bone during osteoarthritis. Cell Death Dis 8: 27–45. https://doi.org/10.1038/cddis.2017.129

Ward JP (2008) Oxygen sensors in context. Biochim Biophys Acta 1777: 1–14. https://doi.org/10.1016/j.bbabio.2007.10.010

Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi Y (2013) Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin 34: 747–754. https://doi.org/10.1038/aps.2013.50

Widmaier M, Rognoni E, Radovanac K, Azimifar SB, Fässler R (2012) Integrin-linked kinase at a glance. J Cell Sci 125: 1839–1843. https://doi.org/10.1242/jcs.093864

Willis GR, Fernandez-Gonzalez A, Anastas J, Vitali SH, Liu X, Ericsson M, Kwong A, Mitsialis SA, Kourembanas S (2018) Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am J Respir Crit Care Med 197: 104–116. https://doi.org/10.1164/rccm.201705-0925OC

Wynn RF, Hart CA, Corradi-Perini C, O’Neill L, Evans CA, Wraith JE, Fairbairn LJ, Bellantuono I (2004) A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood 104: 2643–2645. https://doi.org/10.1182/blood-2004-02-0526

Yeo RW, Lai RC, Zhang B, Tan SS, Yin Y, The BJ, Lim SK (2013) Mesenchymal stem cell: an efficient mass producer of exosomes for drug delivery. Adv Drug Deliv Rev 65: 336–341. https://doi.org/10.1016/j.addr.2012.07.001

Yoon D, Kim H, Lee E, Park MH, Chung S, Jeon H, Ahn CH, Lee K (2016) Study on chemotaxis and chemokinesis of bone marrow-derived mesenchymal stem cells in hydrogel-based 3D microfluidic devices. Biomater Res 20: 25–39. https://doi.org/10.1186/s40824-016-0070-6.

Yun SP, Lee MY, Ryu JM, Song CH, Han HJ (2009) Role of HIF-1alpha and VEGF in human mesenchymal stem cell proliferation by 17beta-estradiol: Involvement of PKC, PI3K/Akt, and MAPKs. Am J Physiol Cell Physiol 296: C317–C326. https://doi.org/10.1152/ajpcell.00415.2008

Zhang L, Zhou Y, Sun XY, Zhou J, Yang PS (2017) CXCL12 overexpression promotes the angiogenesis potential of periodontal ligament stem cells. Sci Rep 7: 102–115. https://doi.org/10.1038/s41598-017-10971-1

Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH (2001) Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 7: 211–228. https://doi.org/10.1089/107632701300062859

Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, Moss J, Burger JA, Maini RN (2000) Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res 2: 477–488. https://doi.org/10.1186/ar130